Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 609(7929): 975-985, 2022 09.
Article in English | MEDLINE | ID: mdl-36104561

ABSTRACT

Understanding cell state transitions and purposefully controlling them is a longstanding challenge in biology. Here we present cell state transition assessment and regulation (cSTAR), an approach for mapping cell states, modelling transitions between them and predicting targeted interventions to convert cell fate decisions. cSTAR uses omics data as input, classifies cell states, and develops a workflow that transforms the input data into mechanistic models that identify a core signalling network, which controls cell fate transitions by influencing whole-cell networks. By integrating signalling and phenotypic data, cSTAR models how cells manoeuvre in Waddington's landscape1 and make decisions about which cell fate to adopt. Notably, cSTAR devises interventions to control the movement of cells in Waddington's landscape. Testing cSTAR in a cellular model of differentiation and proliferation shows a high correlation between quantitative predictions and experimental data. Applying cSTAR to different types of perturbation and omics datasets, including single-cell data, demonstrates its flexibility and scalability and provides new biological insights. The ability of cSTAR to identify targeted perturbations that interconvert cell fates will enable designer approaches for manipulating cellular development pathways and mechanistically underpinned therapeutic interventions.


Subject(s)
Cell Differentiation , Models, Biological , Signal Transduction , Cell Proliferation , Datasets as Topic , Phenotype , Single-Cell Analysis , Workflow
2.
Int J Mol Sci ; 24(14)2023 Jul 23.
Article in English | MEDLINE | ID: mdl-37511580

ABSTRACT

Kinase Suppressor of RAS 1 (KSR1) is a scaffolding protein for the RAS-RAF-MEK-ERK pathway, which is one of the most frequently altered pathways in human cancers. Previous results have shown that KSR1 has a critical role in mutant RAS-mediated transformation. Here, we examined the role of KSR1 in mutant BRAF transformation. We used CRISPR/Cas9 to knock out KSR1 in a BRAFV600E-transformed melanoma cell line. KSR1 loss produced a complex phenotype characterised by impaired proliferation, cell cycle defects, decreased transformation, decreased invasive migration, increased cellular senescence, and increased apoptosis. To decipher this phenotype, we used a combination of proteomic ERK substrate profiling, global protein expression profiling, and biochemical validation assays. The results suggest that KSR1 directs ERK to phosphorylate substrates that have a critical role in ensuring cell survival. The results further indicate that KSR1 loss induces the activation of p38 Mitogen-Activated Protein Kinase (MAPK) and subsequent cell cycle aberrations and senescence. In summary, KSR1 function plays a key role in oncogenic BRAF transformation.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , MAP Kinase Signaling System , Melanoma/genetics , Proteomics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , ras Proteins/metabolism
3.
EMBO Rep ; 19(2): 320-336, 2018 02.
Article in English | MEDLINE | ID: mdl-29263201

ABSTRACT

Altered cell metabolism is a hallmark of cancer, and targeting specific metabolic nodes is considered an attractive strategy for cancer therapy. In this study, we evaluate the effects of metabolic stressors on the deregulated ERK pathway in melanoma cells bearing activating mutations of the NRAS or BRAF oncogenes. We report that metabolic stressors promote the dimerization of KSR proteins with CRAF in NRAS-mutant cells, and with oncogenic BRAF in BRAFV600E-mutant cells, thereby enhancing ERK pathway activation. Despite this similarity, the two genomic subtypes react differently when a higher level of metabolic stress is induced. In NRAS-mutant cells, the ERK pathway is even more stimulated, while it is strongly downregulated in BRAFV600E-mutant cells. We demonstrate that this is caused by the dissociation of mutant BRAF from KSR and is mediated by activated AMPK. Both types of ERK regulation nevertheless lead to cell cycle arrest. Besides studying the effects of the metabolic stressors on ERK pathway activity, we also present data suggesting that for efficient therapies of both genomic melanoma subtypes, specific metabolic targeting is necessary.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinases/metabolism , Protein Multimerization , Stress, Physiological , raf Kinases/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Enzyme Activation , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Glucose/metabolism , Glycolysis , Humans , Melanoma/genetics , Melanoma/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Oxygen Consumption , Protein Kinases/chemistry , Protein Kinases/genetics , Recombinant Fusion Proteins , raf Kinases/chemistry , raf Kinases/genetics
4.
Angew Chem Int Ed Engl ; 55(3): 983-6, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26644280

ABSTRACT

RAF kinases are key players in the MAPK signaling pathway and are important targets for personalized cancer therapy. RAF dimerization is part of the physiological activation mechanism, together with phosphorylation, and is known to convey resistance to RAF inhibitors. Herein, molecular dynamics simulations are used to show that phosphorylation of a key N-terminal acidic (NtA) motif facilitates RAF dimerization by introducing several interprotomer salt bridges between the αC-helix and charged residues upstream of the NtA motif. Additionally, we show that the R-spine of RAF interacts with a conserved Trp residue in the vicinity of the NtA motif, connecting the active sites of two protomers and thereby modulating the cooperative interactions in the RAF dimer. Our findings provide a first structure-based mechanism for the auto-transactivation of RAF and could be generally applicable to other kinases, opening new pathways for overcoming dimerization-related drug resistance.


Subject(s)
Transcriptional Activation , raf Kinases/metabolism , Dimerization , Phosphorylation , Protein Conformation , raf Kinases/chemistry
5.
Sci Data ; 10(1): 203, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37045861

ABSTRACT

RAF kinases play major roles in cancer. BRAFV600E mutants drive ~6% of human cancers. Potent kinase inhibitors exist but show variable effects in different cancer types, sometimes even inducing paradoxical RAF kinase activation. Both paradoxical activation and drug resistance are frequently due to enhanced dimerization between RAF1 and BRAF, which maintains or restores the activity of the downstream MEK-ERK pathway. Here, using quantitative proteomics we mapped the interactomes of RAF1 monomers, RAF1-BRAF and RAF1-BRAFV600E dimers identifying and quantifying >1,000 proteins. In addition, we examined the effects of vemurafenib and sorafenib, two different types of clinically used RAF inhibitors. Using regression analysis to compare different conditions we found a large overlapping core interactome but also distinct condition specific differences. Given that RAF proteins have kinase independent functions such dynamic interactome changes could contribute to their functional diversification. Analysing this dataset may provide a deeper understanding of RAF signalling and mechanisms of resistance to RAF inhibitors.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction , Vemurafenib , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Proteome
6.
Biomolecules ; 13(8)2023 08 02.
Article in English | MEDLINE | ID: mdl-37627277

ABSTRACT

Cancer cells often adapt to targeted therapies, yet the molecular mechanisms underlying adaptive resistance remain only partially understood. Here, we explore a mechanism of RAS/RAF/MEK/ERK (MAPK) pathway reactivation through the upregulation of RAF isoform (RAFs) abundance. Using computational modeling and in vitro experiments, we show that the upregulation of RAFs changes the concentration range of paradoxical pathway activation upon treatment with conformation-specific RAF inhibitors. Additionally, our data indicate that the signaling output upon loss or downregulation of one RAF isoform can be compensated by overexpression of other RAF isoforms. We furthermore demonstrate that, while single RAF inhibitors cannot efficiently inhibit ERK reactivation caused by RAF overexpression, a combination of two structurally distinct RAF inhibitors synergizes to robustly suppress pathway reactivation.


Subject(s)
Up-Regulation , Computer Simulation , Down-Regulation , Molecular Conformation , Drug Resistance
7.
Cell Rep ; 35(8): 109157, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34038718

ABSTRACT

Increasing evidence suggests that the reactivation of initially inhibited signaling pathways causes drug resistance. Here, we analyze how network topologies affect signaling responses to drug treatment. Network-dependent drug resistance is commonly attributed to negative and positive feedback loops. However, feedback loops by themselves cannot completely reactivate steady-state signaling. Newly synthesized negative feedback regulators can induce a transient overshoot but cannot fully restore output signaling. Complete signaling reactivation can only occur when at least two routes, an activating and inhibitory, connect an inhibited upstream protein to a downstream output. Irrespective of the network topology, drug-induced overexpression or increase in target dimerization can restore or even paradoxically increase downstream pathway activity. Kinase dimerization cooperates with inhibitor-mediated alleviation of negative feedback. Our findings inform drug development by considering network context and optimizing the design drug combinations. As an example, we predict and experimentally confirm specific combinations of RAF inhibitors that block mutant NRAS signaling.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Humans , Signal Transduction
8.
Nat Commun ; 11(1): 499, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980649

ABSTRACT

Protein-protein-interaction networks (PPINs) organize fundamental biological processes, but how oncogenic mutations impact these interactions and their functions at a network-level scale is poorly understood. Here, we analyze how a common oncogenic KRAS mutation (KRASG13D) affects PPIN structure and function of the Epidermal Growth Factor Receptor (EGFR) network in colorectal cancer (CRC) cells. Mapping >6000 PPIs shows that this network is extensively rewired in cells expressing transforming levels of KRASG13D (mtKRAS). The factors driving PPIN rewiring are multifactorial including changes in protein expression and phosphorylation. Mathematical modelling also suggests that the binding dynamics of low and high affinity KRAS interactors contribute to rewiring. PPIN rewiring substantially alters the composition of protein complexes, signal flow, transcriptional regulation, and cellular phenotype. These changes are validated by targeted and global experimental analysis. Importantly, genetic alterations in the most extensively rewired PPIN nodes occur frequently in CRC and are prognostic of poor patient outcomes.


Subject(s)
Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Mutation/genetics , Protein Interaction Maps , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor , Humans , Phosphorylation , Prognosis , Survival Analysis , bcl-Associated Death Protein/metabolism
9.
Cell Rep ; 24(5): 1316-1329, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067985

ABSTRACT

Cellular p53 protein levels are regulated by a ubiquitination/de-ubiquitination cycle that can target the protein for proteasomal destruction. The ubiquitination reaction is catalyzed by a multitude of ligases, whereas the removal of ubiquitin chains is mediated by two deubiquitinating enzymes (DUBs), USP7 (HAUSP) and USP10. Here, we show that PHD3 hydroxylates p53 at proline 359, a residue that is in the p53-DUB binding domain. Hydroxylation of p53 upon proline 359 regulates its interaction with USP7 and USP10, and its inhibition decreases the association of p53 with USP7/USP10, increases p53 ubiquitination, and rapidly reduces p53 protein levels independently of mRNA expression. Our results show that p53 is a PHD3 substrate and that hydroxylation by PHD3 regulates p53 protein stability through modulation of ubiquitination.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Binding Sites , HEK293 Cells , Humans , Protein Binding , Protein Stability , Tumor Suppressor Protein p53/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7/metabolism
10.
Cell Syst ; 7(2): 161-179.e14, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30007540

ABSTRACT

Clinically used RAF inhibitors are ineffective in RAS mutant tumors because they enhance homo- and heterodimerization of RAF kinases, leading to paradoxical activation of ERK signaling. Overcoming enhanced RAF dimerization and the resulting resistance is a challenge for drug design. Combining multiple inhibitors could be more effective, but it is unclear how the best combinations can be chosen. We built a next-generation mechanistic dynamic model to analyze combinations of structurally different RAF inhibitors, which can efficiently suppress MEK/ERK signaling. This rule-based model of the RAS/ERK pathway integrates thermodynamics and kinetics of drug-protein interactions, structural elements, posttranslational modifications, and cell mutational status as model rules to predict RAF inhibitor combinations for inhibiting ERK activity in oncogenic RAS and/or BRAFV600E backgrounds. Predicted synergistic inhibition of ERK signaling was corroborated by experiments in mutant NRAS, HRAS, and BRAFV600E cells, and inhibition of oncogenic RAS signaling was associated with reduced cell proliferation and colony formation.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , ras Proteins/metabolism , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Molecular Docking Simulation , Mutation/drug effects , Neoplasms/genetics , Neoplasms/metabolism , Protein Multimerization/drug effects , Thermodynamics , raf Kinases/chemistry , raf Kinases/metabolism , ras Proteins/genetics
11.
PLoS One ; 12(2): e0171435, 2017.
Article in English | MEDLINE | ID: mdl-28192450

ABSTRACT

We have developed a novel analysis method that can interrogate the authenticity of biological samples used for generation of transcriptome profiles in public data repositories. The method uses RNA sequencing information to reveal mutations in expressed transcripts and subsequently confirms the identity of analysed cells by comparison with publicly available cell-specific mutational profiles. Cell lines constitute key model systems widely used within cancer research, but their identity needs to be confirmed in order to minimise the influence of cell contaminations and genetic drift on the analysis. Using both public and novel data, we demonstrate the use of RNA-sequencing data analysis for cell line authentication by examining the validity of COLO205, DLD1, HCT15, HCT116, HKE3, HT29 and RKO colorectal cancer cell lines. We successfully authenticate the studied cell lines and validate previous reports indicating that DLD1 and HCT15 are synonymous. We also show that the analysed HKE3 cells harbour an unexpected KRAS-G13D mutation and confirm that this cell line is a genuine KRAS dosage mutant, rather than a true isogenic derivative of HCT116 expressing only the wild type KRAS. This authentication method could be used to revisit the numerous cell line based RNA sequencing experiments available in public data repositories, analyse new experiments where whole genome sequencing is not available, as well as facilitate comparisons of data from different experiments, platforms and laboratories.


Subject(s)
DNA Mutational Analysis/methods , Mutation , Sequence Analysis, RNA/methods , Transcriptome/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , HCT116 Cells , HT29 Cells , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Reproducibility of Results
12.
Cell Signal ; 28(10): 1451-62, 2016 10.
Article in English | MEDLINE | ID: mdl-27368419

ABSTRACT

RAF family kinases are central components of the Ras-RAF-MEK-ERK cascade. Dimerization is a key mechanism of RAF activation in response to physiological, pathological and pharmacological signals. It is mediated by a dimer interface region in the RAF kinase domain that is also conserved in KSR, a scaffolding protein that binds RAF, MEK and ERK. The regulation of RAF dimerization is incompletely understood. Especially little is known about the molecular mechanism involved in the selection of the dimerization partner. Previously, we reported that Ras-dependent binding of the tumour suppressor DiRas3 to C-RAF inhibits the C-RAF:B-RAF heterodimerization. Here we show that DiRas3 binds to KSR1 independently of its interaction with activated Ras and RAF. Our data also suggest that depending on the local stoichiometry between DiRas3 and oncogenic Ras, DiRas3 can either enhance homodimerization of KSR1 or recruit KSR1 to the Ras:C-RAF complex and thereby reduce the availability of C-RAF for binding to B-RAF. This mechanism, which is shared between A-RAF and C-RAF, may be involved in the regulation of Ras12V-induced cell transformation by DiRas3.


Subject(s)
Multiprotein Complexes/metabolism , Protein Kinases/metabolism , Protein Multimerization , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Intracellular Space/metabolism , Mice , NIH 3T3 Cells , Protein Binding , Protein Stability , Proto-Oncogene Proteins p21(ras)/metabolism , Subcellular Fractions/metabolism
13.
Curr Opin Struct Biol ; 41: 151-158, 2016 12.
Article in English | MEDLINE | ID: mdl-27521656

ABSTRACT

The RAS/RAF/MEK/MAPK kinase pathway has been extensively studied for more than 25 years, yet we continue to be puzzled by its intricate dynamic control and plasticity. Different spatiotemporal MAPK dynamics bring about distinct cell fate decisions in normal vs cancer cells and developing organisms. Recent modelling and experimental studies provided novel insights in the versatile MAPK dynamics concerted by a plethora of feedforward/feedback regulations and crosstalk on multiple timescales. Multiple cancer types and various developmental disorders arise from persistent alterations of the MAPK dynamics caused by RAS/RAF/MEK mutations. While a key role of the MAPK pathway in multiple diseases made the development of novel RAF/MEK inhibitors a hot topic of drug development, these drugs have unexpected side-effects and resistance inevitably occurs. We review how RAF dimerization conveys drug resistance and recent breakthroughs to overcome this resistance.


Subject(s)
Drug Resistance, Neoplasm , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/pathology , Animals , Carcinogenesis/drug effects , Humans , Mitogen-Activated Protein Kinases/chemistry , Neoplasms/drug therapy , Protein Multimerization/drug effects
14.
Cell Rep ; 14(11): 2745-60, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26972000

ABSTRACT

Amino acid hydroxylation is a post-translational modification that regulates intra- and inter-molecular protein-protein interactions. The modifications are regulated by a family of 2-oxoglutarate- (2OG) dependent enzymes and, although the biochemistry is well understood, until now only a few substrates have been described for these enzymes. Using quantitative interaction proteomics, we screened for substrates of the proline hydroxylase PHD3 and the asparagine hydroxylase FIH, which regulate the HIF-mediated hypoxic response. We were able to identify hundreds of potential substrates. Enrichment analysis revealed that the potential substrates of both hydroxylases cluster in the same pathways but frequently modify different nodes of signaling networks. We confirm that two proteins identified in our screen, MAPK6 (Erk3) and RIPK4, are indeed hydroxylated in a FIH- or PHD3-dependent mechanism. We further determined that FIH-dependent hydroxylation regulates RIPK4-dependent Wnt signaling, and that PHD3-dependent hydroxylation of MAPK6 protects the protein from proteasomal degradation.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Amino Acids, Dicarboxylic/chemistry , Chromatography, High Pressure Liquid , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Immunoblotting , Immunoprecipitation , Mitogen-Activated Protein Kinase 6/antagonists & inhibitors , Mitogen-Activated Protein Kinase 6/metabolism , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Peptides/analysis , Peptides/chemistry , Protein Interaction Maps , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Ubiquitination
15.
Cell Signal ; 28(9): 1432-1439, 2016 09.
Article in English | MEDLINE | ID: mdl-27345148

ABSTRACT

The BRAF proto-oncogene serine/threonine-protein kinase, known as BRAF, belongs to the RAF kinase family. It regulates the MAPK/ERK signalling pathway affecting several cellular processes such as growth, survival, differentiation, and cellular transformation. BRAF is mutated in ~8% of all human cancers with the V600E mutation constituting ~90% of mutations. Here, we have used quantitative mass spectrometry to map and compare phosphorylation site patterns between BRAF and BRAF V600E. We identified sites that are shared as well as several quantitative differences in phosphorylation abundance. The highest difference is phosphorylation of S614 in the activation loop which is ~5fold enhanced in BRAF V600E. Mutation of S614 increases the kinase activity of both BRAF and BRAF V600E and the transforming ability of BRAF V600E. The phosphorylation of S614 is mitogen inducible and the result of autophosphorylation. These data suggest that phosphorylation at this site is inhibitory, and part of the physiological shut-down mechanism of BRAF signalling.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Phosphoserine/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation/genetics , Phosphorylation , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/chemistry , Rats
16.
Sci Signal ; 8(408): ra130, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26696630

ABSTRACT

Signaling pathways control cell fate decisions that ultimately determine the behavior of cancer cells. Therefore, the dynamics of pathway activity may contain prognostically relevant information different from that contained in the static nature of other types of biomarkers. To investigate this hypothesis, we characterized the network that regulated stress signaling by the c-Jun N-terminal kinase (JNK) pathway in neuroblastoma cells. We generated an experimentally calibrated and validated computational model of this network and used the model to extract prognostic information from neuroblastoma patient-specific simulations of JNK activation. Switch-like JNK activation mediates cell death by apoptosis. An inability to initiate switch-like JNK activation in the simulations was significantly associated with poor overall survival for patients with neuroblastoma with or without MYCN amplification, indicating that patient-specific simulations of JNK activation could stratify patients. Furthermore, our analysis demonstrated that extracting information about a signaling pathway to develop a prognostically useful model requires understanding of not only components and disease-associated changes in the abundance or activity of the components but also how those changes affect pathway dynamics.


Subject(s)
Biomarkers, Tumor/metabolism , MAP Kinase Kinase 4/metabolism , Models, Biological , Neuroblastoma/metabolism , Neuroblastoma/mortality , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Signal Transduction , Adolescent , Animals , Cell Line, Tumor , Child , Child, Preschool , Disease-Free Survival , Female , Follow-Up Studies , Humans , Infant , Male , N-Myc Proto-Oncogene Protein , Neoplasms, Experimental/metabolism , Predictive Value of Tests , Survival Rate , Zebrafish/metabolism , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL